This application is the U.S. National Phase of International Patent Application Serial No. PCT/EP2014/056380, filed on Mar. 28, 2014, which claims priority to European Patent Application No. EP 13305404.9, filed on Mar. 28, 2013, both of which applications are herein incorporated by reference in their entirety.
BACKGROUND OF THE INVENTION
Like their human counterparts, cats that live in developed countries have seen their life expectancy consistently prolonged. Therefore, the global burden of cancers continues to increase largely because of the aging and growing of the cat population.
Cancer incidence rate is estimated to 77 per 10,000 cats. Lymphomas and tumors of the sub-cutaneous tissues, and especially the complex feline fibrosarcoma, are the most frequent of the feline cancerous diseases (Vascellari et al. 2009).
The panel of treatments available against veterinary cancer is substantially reduced compared with those available in human oncology.
Surgery remains the best way to treat animal tumors. This method presents the advantage of being accessible for many veterinarians, and, in many cases, it can be curative. However, to be curative, surgery must be bold and in some cases the tumor is too large, too dispersed or just not accessible enough to be entirely removed. If not totally curative, surgery can still be a palliative solution to improve animal's comfort and prolonged its life expectancy.
Radiotherapy is another important means to treat certain types of cancers in the veterinary field. It is of particular interest for tumors which are hardly accessible for surgery like cerebral tumors. Furthermore, recent studies in humans have demonstrated that ionizing radiation (IR) could act as an immunomodulator by inducing substantial changes in the tumor microenvironment, including triggering an inflammatory process. Furthermore, the cost and the availability of the material make access to radiation therapy complicated for companion animals.
Chemotherapy is more and more used in animal oncology (Marconato 2011). Taking advantages of medical advances in human cancer therapy, there are more and more molecules available like vincristine, cyclophosphamide, carboplatin or cisplatin, to treat companion animals. In the veterinary field, anticancer drugs are particularly used in the treatment of tumors derived from hematopoietic tissue (lymphomas, leukemias). For example the CHOP protocol, combining cyclophosphamide, doxorubicin, vincristine and prednisone is currently used in the treatment of numerous lymphomas (Chun 2009). Chemotherapeutic agents can be particularly efficient in prolonging the life span of a cancerous animal from a few weeks to several months. Interestingly, the side effects dreaded by human patients, such as vomiting, diarrhea, hair loss, are usually less frequent in companion animals. Unfortunately, most of the time chemotherapy is not curative in pets and the tumor often escapes from treatment.
Therefore, just as in human medicine, targeted therapies are in development in veterinary medicine. Other treatments, including immunotherapies, are under investigation. These immunotherapeutic treatments are all based on the fact that it is possible to activate the immune system of the host against cancer cells.
The relationship between the host immune system and cancer is dynamic and complex. Each type of tumor cells harbors a multitude of somatic mutations and epigenetically deregulated genes, the products of which are potentially recognizable as foreign antigens by immune cells (MUC-1, β-catenin, telomerase . . . ) (Fridman et al. 2012). Growing tumors contain infiltrating lymphocytes called TILs (Tumor Infiltrating Lymphocytes). These killer cells are often ineffective at tumor elimination in vivo but can exert specific functions in vitro, that is to say outside the immunosuppressive tumor microenvironment (Restifo et al. 2012). This is because the tumor stroma contains many suppressive elements including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDCs); soluble factors such as interleukin 6 (IL-6), IL-10, vascular endothelial growth factor (VEGF), and transforming growth factor beta (TGFβ that down modulate antitumor immunity (Finn 2008, Hanahan and Weinberg 2011). Consequently, the choice of a pertinent tumor associated antigen (TAA) and the bypass of cancer associated immunosuppression are two critical points for a therapeutic vaccine to succeed (Disis et al. 2009).
Recent introduction of active cancer immunotherapy (also referred to cancer vaccines) in the clinical cancer practice emphasizes the role of immune responses in cancer prognosis and has led to a growing interest to extend this approach to several human and companion animal cancers (Dillman 2011, Topalian et al. 2011) (Jourdier et al. 2003).
In this context, there is still a need for an innovative cancer vaccine strategy for cats, which would overcome the challenge of breaking tolerance and inducing an immune response in the animal.
SUMMARY OF THE INVENTION
The inventors now propose a cancer vaccine strategy for cats, based on the telomerase reverse transcriptase (TERT).
A subject of the invention is thus an immunogenic composition comprising a nucleic acid that comprises a sequence encoding (i) a cat TERT deprived of telomerase catalytic activity, or (ii) a fragment thereof. The nucleic acid is preferably DNA, preferably in form of a plasmid.
In a preferred embodiment, the nucleic acid that comprises a sequence encoding a cat telomerase reverse transcriptase (TERT) deprived of telomerase catalytic activity, wherein the sequence encoding catTERT is further deprived of a nucleolar localization signal.
In a particular embodiment, the nucleic acid further comprises a non-cat TERT antigenic fragment.
A further subject of the invention is a nucleic acid that comprises a sequence encoding (i) a cat TERT deprived of telomerase catalytic activity, or (ii) a fragment thereof, and optionally further comprises a non-cat TERT antigenic fragment.
The immunogenic composition or the nucleic acid is useful in triggering an immune response in a cat, against cells that overexpress telomerase, such as dysplasia cells, tumor cells, or cells infected by an oncovirus.
The immunogenic composition or the nucleic acid is thus particularly useful in treating a tumor in a cat, preferably by intradermal or intramuscular route.
Such treatment can be referred to as an active immunotherapy or a therapeutic vaccination, as it triggers an immune response against the tumor, especially a cytotoxic CD8 T cell response, along with a specific CD4 T cell response.
The invention makes it possible to induce dTERT specific responses in cats with neoplasias and so can be used for immunotherapeutic treatments of the neoplasias in a clinical setting.
The invention is also useful to induce dTERT specific responses in healthy cats that could be at risk for cancer, e.g. by genetic predisposition, or in healthy cats from a certain age (e.g. of 12 years or more, preferably more than 14 years old) so as to prevent the onset of cancer.
Generally speaking, the treatment of the invention may induce long term immune memory responses in healthy dogs, dogs at risk of developing a cancer and those presenting a cancer.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1A shows pUF2 nucleotide sequence (SEQ ID NO: 1) and corresponding amino acid sequence comprising cat TERT amino acid sequence. (SEQ ID NO: 2).
The plasmid pUF2 encodes a cat TERT (cTERT) protein comprising about 95% from the cat TERT and about 5% from the dog TERT sequence. Exon 1 encoding the extreme amino terminus of the cat telomerase genes remains unknown. It is estimated that 47 amino acids (141 bases) are missing. The nucleotide sequence encoding 3 key amino acids in the catalytic site of the protein have been deleted (VDD). Moreover, the sequence controlling the importation into the nucleoli (Nucleolar addressing signal) has been deleted (nucleotide sequence encoding 47 first Amino Acids in the N ter sequence of cTERT protein). The DNA sequence encoding the human ubiquitin has been added upstream the cTERT sequence. The presence of the ubiquitin protein enhances the addressing of the cTERT protein to the proteasome and increases class I presentation of derived peptides. However, as the human and cat ubiquitin sequences are identical at the protein level, there is no biological incompatibility. Downstream the cTERT sequence, the sequence of the V5 peptide of the flu was inserted to facilitate the detection of the protein
Nucleotides 1-6 HindIII restriction site for subcloning
Nucleotides 13-240 ubiquitin
Nucleotides 241-438 dog TERT (5.5% of TERT sequences)
Nucleotides 439-3444 cat TERT Nucleotides 3517-3558 SV5 V5 tag
Nucleotides 3586-3588 two stop codons
Nucleotides 3495-3500 Xba1 restriction site for subcloning
Nucleotides 2655-2656 inactivating deletion of 9 bp encoding VDD residues
FIG. 1B shows pCDT nucleotide sequence (SEQ ID NO: 3) and corresponding amino acid sequence containing cat/dog hybrid TERT amino acid sequence (SEQ ID NO: 4).
The plasmid pCDT encode the cat/dog hybrid TERT (hyTERT) comprising 54.4% from the cat TERT and 35.9% from the dog TERT sequence. The nucleotide sequence encoding 3 key amino acids in the catalytic site of the protein have been deleted (VDD). Moreover, the sequence controlling the importation into the nucleoli (Nucleolar addressing signal) has been depleted (nucleotide sequence encoding 45 first Amino Acids in the Nterm sequence of hyTERT protein). The DNA sequence encoding the human ubiquitin has been added upstream the hyTERT sequence. The presence of the ubiquitin protein enhances the addressing of the hyTERT protein to the proteasome and increases class I presentation of the derived peptides. Downstream the hyTERT sequence, the sequence of the V5 peptide of the flu was inserted to facilitate the detection of the protein.
Nucleotides 1-6 HindIII restriction site for subcloning
Nucleotides 13-240 ubiquitin
Nucleotides 241-1413 dog TERT (35.9% of TERT sequences)
Nucleotides 1414-3297 cat TERT (54.4% of TERT sequences)
Nucleotides 3298-3456 dog TERT last exon
Nucleotides 3457-3510 influenza A2 epitope
Nucleotides 3511-3552 SV5 V5 tag
Nucleotides 2667-2668 inactivating deletion of 9 bp encoding VDD residues
Nucleotides 3553-3558 two stop codons
Nucleotides 3559-3564 Xba1 restriction site for subcloning
FIG. 1C shows a simplified map of pcDNA3.1 expression plasmid into which the cat/dog hybrid TERT nucleic acid sequence was cloned.
FIG. 2 shows that pDNA constructs are safe (Trapeze), (A) Lysates obtained from CrFK cells transfected with hTERT (human telomerase fully active), pCDT or pUF2 plasmids were analyzed for telomerase activity by the TRAP assay. The level of telomerase activity is shown as relative telomerase activity compared with that of control template measured in each kit. All samples at 2.1 μg protein concentration were measured in triplicate, error bars are standard error of the mean (SEM) (**P=0.0020, hTERT vs pUF2 unpaired t test)
FIGS. 3A and 3B show specific IFNγ+ CD8 and CD4 T-cell responses against H2 restricted hyTERT peptides in mice immunized with pCDT.
Seven week-old female mice were immunized intradermally (ID) or intramuscularly (IM) with either 100 μg pCDT plasmid or PBS at day 0 and boost 14 days later. Ten day post-boost, spleens were harvested. Splenocytes were Ficoll-purified and stimulated in triplicates with 5 μg/mL of relevant peptides for 19 hours. Spots were revealed with a biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution.
(A) Plasmid vaccinated groups were composed of five C57/Bl6 mice, and control groups, of three mice. Splenocytes were stimulated with class I peptides p580, p621 and p987. Results show the frequency of peptide specific IFN-γ producing CD8 T cells.
(B) Plasmid vaccinated groups were composed of 9 Balb/cBy mice immunized IM and 5 ID. Control groups of 8 Balb/cBy mice injected IM and 4 ID. Splenocytes were stimulated with class II peptides p951, p1105, p1106 and p1109. Results show the frequency of peptide specific IFN-γ producing CD4 T cells.
Results are the mean±standard deviation. Mann Whitney non parametric test, * p-value <0.05, **: p-value <0.01.
FIGS. 4A and 4B show a hyTERT specific cytotoxic T-lymphocyte (CTL) response in mice immunized with pCDT plasmid, measurable in vivo by elimination of transferred target cells pulsed with H2 restricted hybrid TERT peptides.
7 week-old C57/Bl6 female mice were immunized ID or IM with 100 μg pCDT plasmid at day 0 and day 14 post-priming. At day 9 post-boost injection, syngeneic splenocytes, pulsed with individual dTERT peptides restricted to H2 (either p987 or p621) or left unpulsed were labeled with carboxyfluorescein-diacetate succinimidyl ester (CFSE) at three different concentrations: high=1 μM (987), medium=0.5 μM (621) and low=0.1 μM (unpulsed). The same number of high, medium or low CFSE labeled cells was transferred IV to vaccinated mice. After 15-18 hours, the disappearance of peptide-pulsed cells was determined by fluorescence-activated cell-sorting analysis in the spleen. The percentage of specific lysis was calculated by comparing the ratio of pulsed to un-pulsed cells in vaccinated versus control mice.
(A) Example of the in vivo CTL assay showing the elimination of target cells pulsed with p621 peptide (High, H) or p987 peptide (Medium, M) in the spleen of a mouse vaccinated ID (left panel) with pCDT. No such disappearing is observed in control mice injected ID with PBS 1× (right panel).
(B) Percentage of specific lysis for each mouse against each individual peptide in the spleen after IM or ID vaccination with pCDT. Horizontal bars show average percentage of lysis per peptide and per immunization route. Standard deviations are also plotted. Representative data from 2 independent experiments (n=10 individual animals/group). Kruskal-Wallis analysis with Dunn's multiple comparison test, * p<0.1, *** p<0.001, ns: not significant. Statistical significance is set at p-value <0.05.
FIGS. 5A and 5B show IFNγ+ specific CD8 and CD4 T-cell responses against H2 restricted cat TERT peptides in mice immunized with pUF2.
Seven week-old female mice were immunized ID or IM with either 100 μg pUF2 plasmid or PBS at day 0 and boost 14 days later. Ten days post boost, spleens were harvested. Splenocytes were Ficoll-purified and stimulated in triplicates with 5 μg/mL of relevant peptides for 19 hours. Spots were revealed with a biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Vaccinated groups were composed of six C57/Bl6 mice, and control groups, of three mice. Splenocytes were stimulated with class I peptides p580, p621 and p987. Results show the frequency of peptide specific IFN-γ producing CD8 T cells. Vaccinated groups were composed of six Balb/cBy mice, and control groups, of three mice. Splenocytes were stimulated with class II peptides p1105 and p1106. Results show the frequency of peptide specific IFN-γ producing CD4 T cells.
Results are the mean±standard deviation. Mann Whitney non parametric test, * p-value <0.05, **: p-value <0.01.
FIGS. 6 A and B show that mice immunized with pUF2 are able to lyse H2 restricted cat TERT peptide-loaded on target cells in vivo
7 week-old C57/Bl6 female mice were immunized ID or IM with 100 μg pCDT plasmid at day 0 and day 14 post-priming. At day 9 post-boost injection, syngeneic splenocytes, pulsed with individual dTERT peptides restricted to H2 (either p987 or p621) or left unpulsed were labeled with carboxyfluorescein-diacetate succinimidyl ester (CFSE) at three different concentrations: high=1 μM (987), medium=0.5 μM (621) and low=0.1 μM (unpulsed). The same number of high, medium or low CFSE labeled cells was transferred IV to vaccinated mice. After 15-18 hours, the disappearance of peptide-pulsed cells was determined by fluorescence-activated cell-sorting analysis in the spleen. The percentage of specific lysis was calculated by comparing the ratio of pulsed to un-pulsed cells in vaccinated versus control mice.
(A) Example of the in vivo CTL assay showing the elimination of target cells pulsed with either p621 or p987 peptides in the spleen of a mouse vaccinated ID (left panel). No such disappearing is observed in control mice (right panel) or in certain mice vaccinated IM (middle panel). H=high, M=Medium, L=Low.
(B) Percentage of specific lysis for each mouse against each individual peptide in the spleen after IM or ID vaccination with pUF2. Horizontal bars show average percentage of lysis per peptide and per immunization route. Standard deviations are also plotted. Representative data from n=5 animals/group. Kruskal-Wallis analysis with Dunn's multiple comparison test, ns: not significant. Statistical significance is set at p-value <0.05.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
The telomerase consists of an RNA template and protein components including a reverse transcriptase, designated “Telomerase Reverse Transcriptase” (TERT), which is the major determinant of telomerase activity. Unless otherwise specified, in the present specification, the term “telomerase” refers to TERT.
In the present invention, the term “cat TERT” refers to the TERT sequence of any domestic cat (also designated as Felis catus or Felis silvestris catus). Partial molecular cloning of the cat TERT gene (237 bp of mRNA) has been reported by Yazawa et al, 2003. The inventors herein provide a longer sequence of Felis catus TERTPartial amino acid sequences of cat TERT are shown as SEQ ID NO:5 and SEQ ID NO:6.
The invention can also make use of non-cat telomerase (TERT) sequence, which can be from any human or non-human mammal, e.g. from dog. The term “dog TERT” refers to the TERT sequence of any domestic dog (also designated Canis familiaris or Canis lupus familiaris).
A dog TERT mRNA sequence is available with NCBI accession number NM_001031630 (XM_545191). Dog TERT amino acid sequence is shown as SEQ ID NO: 9.
The “telomerase catalytic activity” refers to the activity of TERT as a telomerase reverse transcriptase. The term “deprived of telomerase catalytic activity” means that the nucleic acid sequence encodes a mutant TERT, which is inactive.
The term “hybrid” or “chimeric” amino acid or nucleotide sequence means that part of the sequence originates from one animal species and at least another part of the sequence is xenogeneic, i.e. it originates from at least one other animal species.
When referring to a protein, the term “fragment” preferably refers to fragment of at least 10 amino acids, preferably at least 20 amino acids, still preferably at least 30, 40, 50, 60, 70, 80 amino acid fragments.
In the context of the invention, the term “antigenic fragment” refers to an amino acid sequence comprising one or several epitopes that induce T cell response in the animal, preferably cytotoxic T lymphocytes (CTLs). An epitope is a specific site which binds to a T-cell receptor or specific antibody, and typically comprises about 3 amino acid residues to about 30 amino acid residues, preferably 8 or 9 amino acids as far as class I MHC epitopes are concerned, and preferably 11 to 25 amino acids as far as class II MHC epitopes are concerned.
The term “immunogenic” means that the composition or construct to which it refers is capable of inducing an immune response upon administration (preferably in a cat). “Immune response” in a subject refers to the development of a humoral immune response, a cellular immune response, or a humoral and a cellular immune response to an antigen. A “humoral immune response” refers to one that is mediated by antibodies. A “cellular immune response” is one mediated by T-lymphocytes. It includes the production of cytokines, chemokines and similar molecules produced by activated T-cells, white blood cells, or both. Immune responses can be determined using standard immunoassays and neutralization assays for detection of the humoral immune response, which are known in the art. In the context of the invention, the immune response preferably encompasses stimulation or proliferation of cytotoxic CD8 T cells and/or CD4 T cells.
As used herein, the term “treatment” or “therapy” includes curative treatment. More particularly, curative treatment refers to any of the alleviation, amelioration and/or elimination, reduction and/or stabilization (e.g., failure to progress to more advanced stages) of a symptom, as well as delay in progression of the tumor or dysplasia, or of a symptom thereof.
As used herein, the term “prevention” or “preventing” refers to the alleviation, amelioration and/or elimination, reduction and/or stabilization (e.g., failure to progress to more advanced stages) of a prodrome, i.e. any alteration or early symptom (or set of symptoms) that might indicate the start of a disease before specific symptoms occur. A cell that “overexpresses telomerase” refers to a cell in a subject, which either expresses telomerase, e.g. upon mutation or infection, whereas it does usually not, under normal conditions, or to a cell in a subject which expresses a higher level of telomerase (e.g. upon mutation or infection), when compared to normal conditions. Preferably the cell that overexpresses telomerase shows an increase of expression of at least 5%, at least 10%, at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or more.
Nucleic Acid Constructs
It is herein provided a nucleic acid that comprises a sequence encoding (i) a cat telomerase reverse transcriptase (TERT) deprived of telomerase catalytic activity, or (ii) a fragment thereof.
The nucleic acid may be DNA or RNA, but is preferably DNA, still preferably double stranded DNA.
As a first safety key, the TERT sequence is deprived of telomerase catalytic activity. In a preferred embodiment, the sequence that encodes cat TERT contains mutations that provide inactivation of the catalytic activity. The term “mutation” include a substitution of one or several amino acids, a deletion of one or several aminoacids, and/or an insertion of one of several amino acids. Preferably the sequence shows a deletion, preferably a deletion of amino acids VDD, as shown in FIG. 1A or 1B.
As a second safety key, the sequence encoding cat TERT can further be deprived of a nucleolar localization signal. This nucleolar localization signal is correlated with the enzymatic activity of TERT. This signal corresponds to the N-terminal 47 amino acids at the N-terminus of the TERT sequence.
Preferably the sequence encoding cat TERT is deleted of N-terminal 47 amino acids. Cat TERT sequence fragments deleted of amino acids VDD and of the N-terminal nucleolar localization signal are shown as SEQ ID NO:7 and SEQ ID NO:8.
In a particular embodiment, the nucleic acid may encode cat TERT sequence or a fragment thereof only, which preferably corresponds to at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or at least 95% of the cat TERT sequence deleted of the N-terminal 47 amino acids.
Preferably, the nucleic acid encodes a cat TERT sequence comprising, or consisting of, SEQ ID NO: 5, 6, 7 or 8.
The nucleic acid may further encode a non-cat TERT antigenic fragment. This embodiment is preferred, to favor breakage of tolerance towards a self-antigen, and induce an efficient immune response along, with an immune memory response in the cat. The presence of non-cat TERT fragment(s) advantageously engages certain subtypes of CD4+ T cells, providing help for anti-tumor immunity, and reversing potential regulation via the secretion of Th1 cytokines.
The cat and non-cat TERT sequences or fragments thereof are preferably fused, to be expressed as a hybrid or chimeric protein. Alternatively, the cat and non-cat TERT sequences or fragments thereof may be separated, but carried on the same vector, e.g. the same plasmid.
Preferably the non-cat TERT antigenic fragment corresponds to a fragment absent or eliminated from the cat TERT sequence, to the extent it does not complement the loss of catalytic activity or the loss of the nucleolar localization signal.
The cat TERT sequence, or fragment thereof, can represent at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or at least 95% of all TERT sequences in the nucleic acid, plasmid, or other vector.
In a preferred embodiment, the cat TERT sequence or fragment represents at least 90% of the hybrid or chimeric TERT protein.
In another embodiment, the cat TERT sequence or fragment represents at least 60% of the hybrid or chimeric TERT protein.
The non-cat TERT antigenic fragment preferably originates from a dog TERT sequence.
The non-cat TERT antigenic fragment is advantageously processed by dendritic cells, thereby generating T cell help.
In a preferred embodiment, the invention employs a nucleic acid that encodes a protein sequence selected from the group consisting of SEQ ID NO: 2, 4, 5, 6, 7, or 8.
Such nucleic acid may comprise a sequence selected from the group consisting of SEQ ID NO: 1, 3, or nucleotides 241-3444, or 382-3444 or 439-3444 of SEQ ID NO:1, or nucleotides 1408-3297 or 1414-3297 or 241-3456 of SEQ ID NO: 3.
In a particular embodiment, the nucleic acid may further encode a protein which enhances the addressing of the TERT protein to the proteasome and increases class I presentation of the derived peptides. Said protein may be preferably ubiquitin or it may be any chaperon protein, e.g. calreticulin.
Genetic Constructs, Immunogenic Compositions and Administration
Preferably, the nucleic acid is a genetic contrast comprising a polynucleotide sequence as defined herein, and regulatory sequences (such as a suitable promoter(s), enhancer(s), terminator(s), etc.) allowing the expression (e.g. transcription and translation) of the protein product in the host cell or host organism.
The genetic constructs of the invention may be DNA or RNA, and are preferably double-stranded DNA. The genetic constructs of the invention may also be in a form suitable for transformation of the intended host cell or host organism, in a form suitable for integration into the genomic DNA of the intended host cell or in a form suitable for independent replication, maintenance and/or inheritance in the intended host organism. For instance, the genetic constructs of the invention may be in the form of a vector, such as for example a plasmid, cosmid, YAC, a viral vector or transposon. In particular, the vector may be an expression vector, i.e. a vector that can provide for expression in vitro and/or in vivo (e.g. in a suitable host cell, host organism and/or expression system).
In a preferred but non-limiting aspect, a genetic construct of the invention comprises i) at least one nucleic acid of the invention; operably connected to ii) one or more regulatory elements, such as a promoter and optionally a suitable terminator; and optionally also iii) one or more further elements of genetic constructs such as 3′- or 5′-UTR sequences, leader sequences, selection markers, expression markers/reporter genes, and/or elements that may facilitate or increase (the efficiency of) transformation or integration.
In a particular embodiment, the genetic construct can be prepared by digesting the nucleic acid polymer with a restriction endonuclease and cloning into a plasmid containing a promoter such as the SV40 promoter, the cytomegalovirus (CMV) promoter or the Rous sarcoma virus (RSV) promoter. In a preferred embodiment, the TERT nucleic acid sequences are inserted into a pcDNA3.1 expression plasmid (see FIG. 1C) or pcDNA3.1 TOPO-V5. Other vectors include retroviral vectors, lentivirus vectors, adenovirus vectors, vaccinia virus vectors, pox virus vectors and adenovirus-associated vectors.
Compositions can be prepared, comprising said nucleic acid or vector. The compositions are immunogenic. They can comprise a carrier or excipients that are suitable for administration in cats (i.e. non-toxic, and, if necessary, sterile). Such excipients include liquid, semisolid, or solid diluents that serve as pharmaceutical vehicles, isotonic agents, stabilizers, or any adjuvant. Diluents can include water, saline, dextrose, ethanol, glycerol, and the like. Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others. Stabilizers include albumin, among others. Any adjuvant known in the art may be used in the vaccine composition, including oil-based adjuvants such as Freund's Complete Adjuvant and Freund's Incomplete Adjuvant, mycolate-based adjuvants, bacterial lipopolysaccharide (LPS), peptidoglycans, proteoglycans, aluminum hydroxide, saponin, DEAE-dextran, neutral oils (such as miglyol), vegetable oils (such as arachis oil), Pluronic® polyols.
The nucleic acid or composition can be administered directly or they can be packaged in liposomes or coated onto colloidal gold particles prior to administration. Techniques for packaging DNA vaccines into liposomes are known in the art, for example from Murray, 1991. Similarly, techniques for coating naked DNA onto gold particles are taught in Yang, 1992, and techniques for expression of proteins using viral vectors are found in Adolph, 1996.
For genetic immunization, the vaccine compositions are preferably administered intradermally, subcutaneously or intramuscularly by injection or by gas driven particle bombardment, and are delivered in an amount effective to stimulate an immune response in the host organism. In a preferred embodiment of the present invention, administration comprises an electroporation step, also designated herein by the term “electrotransfer”, in addition to the injection step (as described in Mir 2008, Sardesai and Weiner 2011).
The compositions may also be administered ex vivo to blood or bone marrow-derived cells using liposomal transfection, particle bombardment or viral transduction (including co-cultivation techniques). The treated cells are then reintroduced back into the subject to be immunized.
While it will be understood that the amount of material needed will depend on the immunogenicity of each individual construct and cannot be predicted a priori, the process of determining the appropriate dosage for any given construct is straightforward. Specifically, a series of dosages of increasing size, starting at about 5 to 30 μg, or preferably 20-25 μg, up to about 500 μg for instance, is administered to the corresponding species and the resulting immune response is observed, for example by detecting the cellular immune response by an Elispot assay (as described in the experimental section), by detecting CTL response using a chromium release assay or detecting TH (helper T cell) response using a cytokine release assay.
In a preferred embodiment, the vaccination regimen comprises one to three injections, preferably repeated three or four weeks later.
In a particular embodiment, the vaccination schedule can be composed of one or two injections followed three or four weeks later by at least one cycle of three to five injections.
In another embodiment, a primer dose consists of one to three injections, followed by at least a booster dose every year, or every two or years for instance.
Prevention or Treatment of Tumors
The nucleic acid or immunogenic composition as described above is useful in a method for preventing or treating a tumor in a cat.
A method for preventing or treating a tumor in a cat is described, which method comprises administering an effective amount of said nucleic acid or immunogenic composition in a cat in need thereof. Said nucleic acid or immunogenic composition is administered in an amount sufficient to induce an immune response in the cat.
The tumor may be any undesired proliferation of cells, in particular a benign tumor or a malignant tumor, especially a cancer.
The cancer may be at any stage of development, including the metastatic stage. However preferably the cancer has not progressed to metastasis.
In particular the tumor may be selected from the group consisting of a lymphoma or lymphosarcoma (LSA), adenoma, lipoma, myeloproliferative tumor, melanoma, squamous cell carcinoma, mast cell tumor, osteosarcoma, fibrosarcoma, lung tumor, brain tumor, nasal tumor, liver tumor, and mammary tumor.
Lymphoma or lymphosarcoma (LSA) is common among cats with Feline Leukemia Virus (FeLV) infections. LSA affects the intestines and other lymphatic tissues (commonly the abdominal organs).
Adenomas are tumors that affect sebaceous glands predominantly in the limbs, the eyelids and the head. They are also commonly-found in the ears (and ear canals) of cats and may lead to the development of hyperthyroidism.
Lipomas are tumors that occur within the fatty tissues and reside as soft, fluctuant round masses that adhere tightly to surrounding tissue (typically to organs and the membrane linings of body cavities).
Myeloproliferative tumors generally are genetic disorders. It can affect the bone marrow, white blood cells, red blood cells, and platelets.
Melanomas manifest as basal cell tumors. These tumors are usually benign in nature. They are commonly found around the neck, head, ears, and shoulder regions and can be treated through chemotherapy or radiation therapy.
Squamous cell carcinomas affect areas that lack natural pigmentation (oral cavity, tonsils, lips, nose, eyelids, external ear, limbs, toes and nails), or areas that are under constant trauma and irritation. Oral squamous carcinomas are the most common.
Mast cell tumors are either sole or multiple skin nodules that may be ulcerated and pigmented. They can be located on any part of the cat's body.
Osteosarcoma are tumors that mainly affect the joints, bones and lungs.
Fibrosarcomas arise from the fibrous tissues just beneath the skin. Fibrosarcomas generally develop in muscle or in the connective tissue of the body.
Generally speaking, lung tumors, brain tumors, nasal tumors, liver tumors, mammary tumors are encompassed.
In a particular embodiment, the vaccination according to the invention may be combined with conventional therapy, including chemotherapy, radiotherapy or surgery. Combinations with adjuvant immunomodulating molecules such GM-CSF or IL-2 could also be useful.
The Figures and Examples illustrate the invention without limiting its scope.
EXAMPLES
The inventors have constructed DNA vaccines encoding an inactivated form of cat TERT and a cat/dog hybrid TERT (Example 1), and have assessed their functionality, safety and immunogenicity.
They have demonstrated that the plasmids were correctly processed in vitro after transfection in mammalian cells and that the plasmid product of expression (TERT protein) was well expressed. Moreover, no enzymatic activity was detected and TERT proteins were found excluded for the transfected cells nucleoli, which evidences safety of the constructs (Example 2).
Then, the plasmids were found to be immunogenic and to elicit specific efficient CD8 T cells and CD4 T cells in mice (Example 3).
Example 1: Construction of the DNA Plasmids
In all constructs, the TERT sequence is preceded by a DNA sequence encoding the human-ubiquitin. The presence of the Ubiquitin will increase the addressing of the TERT protein to the proteasome and increase the class I presentation pathway of TERT derived peptides. TERT sequence is followed by the sequence of the influenza protein V5 to facilitate future purification or detection of the fusion protein by Western Blot or histochemistry for example.
The DNA sequence coding for the TERT protein has been deleted of 47 Amino-acids in the N-Term region, which encodes the nucleolar importation signal. Moreover, three amino-acids have been removed in the catalytic site of TERT (VDD), to inhibit the protein enzymatic activity. pUF2 encodes 95% of the cat TERT and 5% of the canine TERT sequence (FIG. 1A), pCDT encodes 54.4% of the cat TERT sequence and 35.9% of the dog TERT sequence (FIG. 1B).
All TERT DNA sequences were synthetized from Genecust (Dudelange, Luxembourg). Then they were cloned into the pCDNA3.1 or pcDNA3.1 TOPO-V5 expression plasmid provided by Life technologies SAS (Saint-Aubin, France) using the HindIII and XbaI restriction sites (see FIG. 1C). Plasmids were stored at −20° C., in PBS 1×, at a concentration of 2 mg/mL prior use. The backbone plasmid was used as empty vector for western blot and Trap-Assay experiments. It consists of the pcDNA3.1 backbone plasmid deprived of the transgene protein DNA sequence (TERT).
Example 2: Functionality and Safety of the Plasmids
2.1. Materials and Methods
Cell Culture
The 293T cell line used for transfection assays and immune-fluorescence experiments were kindly provided by Pr Simon Wain-Hobson (Pasteur Institute). CrFK cells were kindly provided by Pr J. Richardson (Ecole Vétérinaire de Maison-Alfort). Cells were grown at 37° C., 5% CO2- in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% heat-inactivated Fetal Calf Serum (FCS), 1% sodium-pyruvate, 1% penicillin-streptomycin pyruvate and 0, 1% β-mercaptoethanol. All components of the culture medium were purchased from Life technologies SAS (Saint-Aubin, France).
Transfection Assays
Transfection of 293T cells were performed with either pCDT or pUF2 plasmids using the JetPRIME® transfection kit (Polyplus-transfection SA, Illkirch, France) according to manufacturer's instruction. In a 6-well plate, 400 000 HeLa cells or 293T cells per well were seeded in 2 mL of DMEM culture medium, and cultured 24 hours at 37° C., 5% CO2 prior transfection. For each well, 2 μg of each plasmid diluted in 200 μL of jetPRIME® buffer, or 200 μL of jetPRIME® buffer only with respectively 4 μL of jetPRIME® agent were drop onto the cells. Transfection medium were removed 4 hours later and replaced by 2 mL of DMEM culture medium. Cells were put at 37° C., 5% CO2 and recovered for analysis 24 hours later.
Western Blots
Transfected 293 T cells were lysed on ice with radioimmunoprecipitation assay (RIPA) lysis buffer (RIPA Buffer, Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) containing protease inhibitors cocktail (Complete EDTA-free, Roche Diagnostic, Indianapolis, USA) for 10-20 minutes. Then, suspension was centrifuged 15 minutes at 14000 rpm at 4° C. in order to remove cellular debris. The supernatants were harvested and the protein concentration was measured using the Bradford method. Protein samples were denatured 5 minutes at 95° C., separated on Nu-PAGE® Novex 4-12% Bis-Tris gels (Invitrogen, Carlsbad, USA) and transferred to PVDF membranes (iBlot® transfer stack, Invitrogen, Carlsbad, USA) using the iBlot® device (Invitrogen, Carlsbad, USA). The membrane was cut approximately at 60 kDa. First, the upper part membrane was probed with an anti-V5 antibody (Invitrogen, Carlsbad, USA) while the other part was probed with an anti-β-actin antibody (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France), then samples were revealed by an ECL (Enhanced chemiluminescence) anti-mouse Horse Radish Peroxidase (HRP) linked antibody (GE Healthcare, Vélizy, France)). Immunoblot signals were reveled using 18×24 films and the corresponding cassette both products purchased from GE healthcare (Buckinghamshire, UK).
Immunofluorescence and Microscopy
293T cells were seeded on 8-well Lab-Tek® chamber slides (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) at 20·103 cells/well in 200 μL of culture medium and incubated overnight at 37° C. The next day, culture medium was discarded. Ten μL of a mix solution containing 1 μg of either pCDT or pUF2 plasmid, 50 μL of OptiMEM (Life technologies SAS, Saint-Aubin, France) and 2.5 μL of Fugene HD (Promega France, Charbonnières-les-bains, France) were added to the corresponding chamber. As control, 20·103 HeLa cells were incubated with the 10 μL of the same mix without plasmid. Chamber slides were left in the incubator for 24 hours. Transfected 293T cells were carefully washed with PBS 1× and 200 μL 2% PFA were added to each well for 10 minutes at +4° C., in order to fix and permeabilize the cells. Then wells were washed two times with PBS 1×0.05% Tween®20 and 293T cells were incubated 30 minutes at room temperature with 200 μL of Blocking solution (0.5% TritonX100; 3% BSA; 10% Goat Serum). Eventually, wells were incubated for 1.5 hours at room temperature with a primary mouse anti-V5 antibody (Life technologies SAS, Saint-Aubin, France) diluted in blocking solution at 1/200, with slight agitation. After three washes in PBS 1×0.05% Tween®20, a secondary goat anti-mouse-Alexa Fluor 488® antibody (Life technologies SAS, Saint-Aubin, France) diluted in blocking solution (1/500) was put in the wells for 45 minutes at room temperature away from light and under slight agitation. Wells were washed three times with PBS 1×0.05% Tween®20 and mounted with the Vectashield® mounting medium containing DAPI (Vector laboratories, Peterborough, UK). Slides were analyzed with a fluorescence microscope (Axio observer Z1, Carl Zeis MicroImaging GmbH, Jena, Germany) equipped with an image processing and analysis system (Axiovision, Carl Zeis MicroImaging GmbH, Jena, Germany).
Trap-Assay
Telomerase activity was measured by the photometric enzyme immunoassay for quantitative determination of telomerase activity, utilizing telomeric repeat amplification protocol (TRAP) (Yang et al. 2002).
CrFK (Crandell Rees Feline Kidney) telomerase-negative cells (Yazawa et al., 2003) were transfected with plasmids encoding pUF2 or pCDT TERT constructs. Briefly, 24 hours after transfection, CrFK cells were harvested by mechanical scraping and then washed twice with 1 mL PBS and pelleted by centrifugation 5 minutes at 3000 g, at 4° C. Telomerase activity was assessed by TRAP-ELISA assay using the TeloTAGGG Telomerase PCR ELISAPLUS kit (Roche Diagnostics, Germany) according to the manufacturer's instructions. The protein concentration in the cell extract was measured by the Bradford method (Bio-Rad Laboratories). Three microliters of the cell extract (equivalent to 2.1, 0.21, 0.021 μg) was incubated in a Polymerase Chain reaction (PCR) mixture provided in the kit. The cycling program was performed with 30 minutes primer elongation at 25° C. and then the mixture was subjected to 30 cycles of PCR consisting of denaturation at 94° C. for 30 sec, annealing at 50° C. for 30 sec, polymerization at 72° C. for 90 sec and final extension at 72° C. for 10 minutes. 2.5 μl of amplification product was used for ELISA according to the manufacturer's instructions. The absorbance at 450 nm (with a reference of 690 nm) of each well was measured using Dynex MRX Revelation and Revelation TC 96 Well Microplate Reader.
Telomerase activity was calculated as suggested in the kit's manual and compared with a control template of 0.1 amol telomeric repeats, representing a relative telomerase activity (RTA) of 100. Inactivated samples and lysis buffer served as negative controls.
2.2. Results
New TERT Encoding Plasmids are Functional In Vitro after Transfection
The functionality of the new plasmid constructs is shown by the presence of the plasmid encoded TERT protein in the total protein lysate of pCDT or pUF2 transfected cells in vitro. The inventors performed western-blot assays on the total protein lysate of 293T cells plasmids transfected with pCDT or pUF2 (24 h after transfection). As the TERT protein sequence encoded by each plasmid was tagged with the V5 protein sequence, anti-V5 antibody coupled with Horse Radish Peroxidase (HRP) was used to reveal the presence of the fusion protein of interest.
A highly positive V5 specific-signal was detected 24 h after transfection in the protein lysate of pCDT or pUF2 transfected cells. The size of the protein band detected corresponds to the different TERT protein encoded by the plasmids which molecular weight is 123 kDa. Moreover no V5 specific signal was detected in untreated or empty plasmid transfected cells. The inventors demonstrated that pUF2 and pCDT plasmids were correctly processed in vitro after transfection in mammalian cells and that the plasmid product of expression (TERT protein) was well expressed.
New TERT Encoding Plasmids Express a Non-Functional Enzyme of which Cellular Expression is Excluded from the Nucleoli after In Vitro Transfection
To test the absence of enzymatic activity, a TRAPeze assay was performed. As illustrated by FIG. 2, protein lysates from pUF2 or pCDT transfected cells do not exhibit any telomerase activity. As a positive control, the protein extracts from 293T cells transfected with the native human TERT were used. Thus the inventors demonstrated that the TERT proteins encoded by either pCDT or pUF2 plasmids do not express any functional enzymatic activity after in vitro transfection.
The inventors have further investigated the intracellular location of the two plasmid products of expression. To this aim, an in vitro immunofluorescence assay was performed. Briefly, 24 h after in-vitro transfection of 293T cells with either pCDT or pUF2, an anti-V5 antibody coupled to an Alexa-Fluor labeled secondary antibody were used to detect the TERT proteins within the cells. The pCDT and pUF2 encoded TERTs were not detected inside the cell nucleoli contrary to what was observed with 293T cells transfected with the plasmid encoding the native human TERT.
To conclude, the inventors demonstrated that after in vitro transfection with either pUF2 and pCDT plasmids, first the TERT protein expression is excluded from the nucleoli and secondly, these products of expression do not exhibit any enzymatic activity. These two criteria establish the safety of the plasmids and favour their use for in vivo vaccination.
Example 3: In Vivo Immune Response
3.1. Materials and Methods
Mice
Female Balb/cBy and C57BL/6J mice (6-8 week old) were purchased from Janvier laboratories (Saint-Berthevin, France). Animals were housed at the Specific Pathogen Free animal facility of the Pasteur Institute. Mice were anesthetized prior to intradermal (ID) or intramuscular (IM) immunizations, with a mix solution of xylazine 2% (Rompun, Bayer Santé, Loos, France) and Ketamine 8% (Imalgen 1000, Merial, Lyon, France) in Phosphate Buffer Saline 1× (PBS 1×, Life technologies SAS, Saint-Aubin, France), according to individual animal weight and duration of anesthesia (intraperitoneal route). All animals were handled in strict accordance with good animal practice and complied with local animal experimentation and ethics committee guidelines of the Pasteur Institute of Paris.
H2 Restricted Peptides
TERT peptides used in mouse studies (IFNγ ELIspot) were predicted by in-silico epitope prediction in order to bind mouse class I MHC, H2Kb, H2Db or mouse class II H2-IAd using four algorithms available online:
Syfpeithi (http://www.syfpeithi.de/), Bimas (http://www-bimas.cit.nih.gov/), NetMHCpan and SMM (http://tools.immuneepitope.org/main/).
All synthetic peptides were purchased lyophilized (>90% purity) from Proimmune (Oxford, United Kingdom). Lyophilized peptides were dissolved in sterile water at 2 mg/mL and stored in 35 μL aliquots at −20° C. prior use. Details of peptides sequence and H2 restriction is shown in table 1.
TABLE 1 |
|
H2 restricted peptides sequences determined |
by in silico prediction algorithms |
|
H2Db restricted TERT peptides |
621-629 (RPIVNMDYI) | 621 | SEQ ID NO: 10 |
580-589 (RQLFNSVHL) | 580 | SEQ ID NO: 11 |
987-996 (TVYMNVYKI) | 987 | SEQ ID NO: 12 |
|
H2-IAd restricted TERT peptides |
1106-1121 (CLLGPLRAAKAHLSR) | 1106 | SEQ ID NO: 13 |
1105-1120 (RCLLGPLRAAKAHLS) | 1105 | SEQ ID NO: 14 |
951-966 (YSSYAQTSIRSSLTF) | 951 | SEQ ID NO: 15 |
1109-1124 (GPLRAAKAHLSRQLP) | 1109 | SEQ ID NO: 16 |
|
Mice Immunization and In Vivo Electroporation
Intradermal (ID) immunization was performed on the lower part of the flank with Insulin specific needles (U-100, 29G×½″-0.33×12 mm, Terumo, Belgium) after shaving. No erythema was observed after shaving, during and after immunization procedure. Intramuscular immunization (IM) was performed in the anterior tibialis cranialis muscle, also using Insulin specific needles U-100. Each animal received a priming dose of either pCDT or pUF2, independently of vaccine route, corresponding to 100 μg of DNA. All animals were boosted at day 14 post-prime using the same amount of plasmid and the same route of immunization. Directly after ID vaccination, invasive needle electrodes (6×4×2, 47-0050, BTX, USA) are inserted into the skin so that the injection site is placed between the two needle rows (the two needle rows are 0.4 cm apart). Two pulses of different voltages were applied (HV-LV): HV=1125V/cm (2 pulses, 50 μs-0.2 μs pulse interval) and LV=250V/cm (8 pulses, 100V-10 ms-20 ms pulse interval). Immediately after IM immunization the muscle injection site was covered with ultrasonic gel (Labo FH, blue contact gel, NM Medical, France) and surrounded by tweezers electrodes (0.5 cm apart, tweezertrode 7 mm, BTXI45-0488, USA) and voltage was applied using the same parameters than for skin electroporation. The Agilepulse® in vivo system electroporator was used for all experiments (BTX, USA).
For each route of immunization (IM, ID) control mice were treated with the same procedures using the same volume of PBS 1×.
Elispot Assay
Briefly, PVDF microplates (IFN-γ Elispot kit, Diaclone, Abcyss, France, 10×96 tests, ref. 862.031.010P) were coated overnight with capture antibody (anti-mouse IFN-γ) and blocked with PBS 2% milk. Spleens from pDNA-immunized mice were mashed and cell suspensions were filtered through a 70-mm nylon mesh (Cell Strainer, BD Biosciences, France). Ficoll-purified splenocytes (Lymphocyte Separation Medium, Eurobio, France) were numerated using the Cellometer® Auto T4 Plus counter (Ozyme, France) and added to the plates in triplicates at 2×105 or 4×105 cells/well and stimulated with 5 μg/ml of cTERT or hyTERT relevant peptides or Concanavalin A (10 μg/ml), or mock stimulated with serum free culture medium. After 19 hours, spots were revealed with the biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Spots were counted using the Immunospot ELIspot counter and software (CTL, Germany).
In Vivo Cytotoxicity Assay
Briefly, for target cell preparation, splenocytes from naïve C57/Bl6 mice were labeled in PBS 1× containing high (5 μM), medium (1 μM) or low (0.2 μM) concentrations of CFSE (Vybrant CFDA-SE cell-tracer kit; Life technologies SAS, Saint-Aubin, France). Splenocytes labeled with 5 and 1 μM CFSE were pulsed with 2 different H2 peptides at 5 μg/ml for 1 hour and 30 minutes at room temperature. Peptides 987 and 621 were used for pulsing respectively CFSE high and medium labeled naïve splenocytes. CFSE low labeled splenocytes were left unpulsed. Each mouse previously immunized with either pCDT or pUF2 received at day 10 post-boost injection 107 CFSE-labeled cells of a mix containing an equal number of cells from each fraction, through the retro-orbital vein. After 15-18 hours, single-cell suspensions from spleens were analyzed by flow cytometry MACSQUANT® cytometer (Miltenyii, Germany).
The disappearance of peptide-pulsed cells was determined by comparing the ratio of pulsed (high/medium CFSE fluorescence intensity) to unpulsed (low CFSE fluorescence intensity) populations in pDNA immunized mice versus control (PBS 1× injected) mice. The percentage of specific killing per test animal was established according to the following calculation:
[1−[mean(CFSElowPBS/CFSEhigh/mediumPBS)/(CFSElowpDNA/CFSEhigh/mediumpDNA)]]×100.
Statistical Analysis and Data Handling
Prism-5 software was used for data handling, analysis and graphic representations. Data are represented as the mean±standard deviation. For statistical analyses of ELIspot assays we used a Mann Whitney non parametric test, and a Kruskal-Wallis analysis with Dunn's multiple comparison test for in vivo cytotoxicity assay. Significance was set at p-value <0.05.
3.2. Results
pCDT Induces a Strong Cytotoxic CD8 T Cell Response Along with a Specific CD4 T Cell Response after ID or IM Immunization and Electroporation in Mice
In light of the importance of cytotoxic CD8 T cells in antitumor immune responses, the inventors have assessed whether plasmid pCDT was able to promote such an immune response in vivo. Thus, different groups of 9-10 C57-Bl/6 mice were immunized with pCDT by ID or IM injection of the plasmid immediately followed by electroporation. Two weeks later, mice received a boost injection with the same protocol. On day 10 post-boost, mice spleens were harvested and the induced immune response was monitored via an IFN-γ ELISPOT assay using H2 restricted peptides described in Table 1.
Hy-TERT peptides restricted to mouse MHC class I were predicted in silico as described in the material and methods section. As shown in FIG. 3A, a significant augmentation in the frequency of hyTERT specific IFN-γ secreting CD8 T-cells was observed in the spleen of ID and IM vaccinated animals in comparison with control mice. This was observed for 2 out of 3 class I restricted peptides (p621 and p987, p<0.05). No significant difference in the frequency of specific CD8 T cells was observed between IM and ID route for both peptides p921 and p987.
The inventors have further investigated the hyTERT restricted CD4 T cell response. To this aim, 9-10 Balb-C mice were immunized with pCDT by ID or IM injection immediately followed by electroporation and the CD4 specific T cell response was monitored in the spleen as described before using hyTERT IAd restricted peptides (in silico prediction). Balb-C mice were chosen because this mouse strain is known to develop good CD4 T cell responses. As shown in FIG. 3B, when performing the IFN-γ ELISPOT assay, a significant augmentation in the frequency of hyTERT specific IFN-γ secreting CD4 T-cells was observed in the spleen of ID and IM vaccinated Balb/C mice in comparison with control mice injected with PBS 1×. This was observed for 2 out of 3 class I restricted peptides (p1106 and p1105, with respectively for p1106 p<0.05 for ID route and p<0.001 for IM route and for 1105 the difference was not significant for ID route and p<0.01 for IM route). No significant difference in the frequency of specific CD4 T cells was observed between IM and ID route for both peptides p1105 and p1106.
Thus, pCDT construct is able to promote the expansion of hyTERT specific CD8 and CD4 T-cells in mice. The inventors next wanted to show that hyTERT specific CD8 T-cells exhibit a functional cytotoxic activity in vivo, which will be necessary to destroy tumor cells. In order to measure the in vivo cytolytic strength of the CD8+ T-cell response elicited by pCDT immunization, the inventors performed an in vivo cytotoxicity test using carboxyfluorescein-diacetate succinimidyl ester (CFSE)-labelled, peptide-pulsed splenocytes as target cells. 7 week old C57/Bl6 mice which received a prime and boost vaccination with pCDT via the ID or IM route as described before or mock-immunized with phosphate-buffered saline (PBS) were intravenously injected with 107 target cells. Target cells were splenocytes from naïve congenic mice separately labelled with three different concentrations of CFSE and pulsed with individual peptides (p621 or p987) or left un-pulsed as an internal control. After 15-18 hours, spleen cells were obtained and the disappearance of peptide-pulsed cells in control versus immunized mice was quantified by fluorescence-activated cell sorting.
Results show that mice develop CTLs against the 2 peptides p621 and p987 which were predicted in silico. Peptide 987 gives the strongest in vivo lysis. Results were consistent with the ones from the IFN-γ Elispot assays (FIG. 3A). It is worth mentioning that for p621, the mean percent lysis was slightly superior when pCDT was injected via the ID route (mean ID=7.7% vs mean IM=0.2%), however, no significant difference was observed between the two routes of immunization.
pUF2 Induces a Strong Cytotoxic CD8 T Cell Response Along with a Specific CD4 T Cell Response after ID or IM Immunization and EP in Mice
The inventors have further investigated whether the pUF2 plasmid was able to stimulate the cTERT specific CD8 T cell response in mice. To this aim, different groups of 5 C57-Bl/6 mice were immunized with pUF2 by ID or IM injection immediately followed by electroporation. Two weeks later, mice received a boost injection with the same protocol. On day 10 post-boost, mice spleens were harvested and the induced immune response was monitored via an IFN-γ ELISPOT assay using H2 restricted peptides described in Table 1. cTERT peptides restricted to mouse MHC class I were predicted in silico as described in the material and methods section above. As shown in FIG. 5A, a significant increase in the frequency of cTERT specific IFN-γ secreting CD8 T-cells was observed in the spleen of ID and IM vaccinated animals in comparison with control mice. This was observed for 2 out of 3 class I restricted peptides (p621 and p987, with respectively for p621 p<0.05 for ID route and no significant difference for IM route and for p687, p<0.001 for ID route and p<0.01 for IM route). No significant difference in the frequency of specific CD8 T cells was observed between IM and ID route for both peptides p921 and p987. However, the mean frequency of p987 specific CD8 T cells was slightly higher when mice were injected via the ID route, in comparison with the IM route (mean ID=143.2 vs mean IM=54.2). The inventors have further investigated the cTERT restricted CD4 T cell response. To this aim, 9-10 Balb-C mice were immunized ID or IM with pUF2 immediately followed by electroporation and the CD4 specific T cell response was monitored in the spleen as described before using cTERT IAd restricted peptides (in silico prediction). Balb-C mice were chosen because this mouse strain is known to develop good CD4 T cell responses. As shown in FIG. 3B, when performing the IFN-γ ELISPOT assay, a significant augmentation in the frequency of hyTERT specific IFN-γ secreting CD4 T-cells was observed in the spleen of ID and IM vaccinated Balb-C mice in comparison with control mice injected with PBS 1×. This was observed for the 2 II restricted peptides tested (p1106 and p1105, p<0.01 for ID and IM route). No significant difference in the frequency of specific CD4 T cells was observed between IM and ID route for both peptides p1105 and p1106.
Thus, pUF2 construct is able to promote the expansion of cTERT specific CD8 and CD4 T-cells in mice. We next wanted to show that cTERT specific CD8 T-cells exhibit a functional cytotoxic activity in vivo, which will be necessary to destroy tumor cells. In order to measure the in vivo cytolytic strength of the CD8+ T-cell response elicited by pUF2 immunization, we performed an in vivo cytotoxicity test using carboxyfluorescein-diacetate succinimidyl ester (CFSE)-labelled, peptide-pulsed splenocytes as target cells. 7 week old C57/Bl6 mice which received a prime and boost vaccination with pUF2 via the ID or IM route as described before or mock-immunized with phosphate-buffered saline (PBS) were intravenously injected with 107 target cells. Target cells were splenocytes from naïve congenic mice separately labelled with three different concentrations of CFSE and pulsed with individual peptides (p621 or p987) or left un-pulsed as an internal control. After 15-18 hours, spleen cells were obtained and the disappearance of peptide-pulsed cells in control versus immunized mice was quantified by fluorescence-activated cell sorting.
The inventors observed that mice developed CTLs against the 2 peptides p621 and p987 which had been previously identified in silico. Peptide 621 gives the strongest in vivo lysis. These results were concordant with the ones from the IFN-γ Elispot assays (FIG. 5A). Interestingly, a significant difference was observed between the two routes of immunization for p621. Indeed, for p621, the mean percent lysis was superior when pUF2 was injected via the ID route (mean ID=64.5% vs mean IM=11%). A non-significant difference was observed for p987 (mean ID=35.7% vs mean IM=21.3%). This confirms that the pUF2 ID vaccination would allow generating a stronger and larger CD8 T cell response that the IM route.
REFERENCES
- Adolph, K. 1996 ed. “Viral Genome Methods” CRC Press, Florida de Fornel P, Delisle F, Devauchelle P, Rosenberg D. 2007. Effects of radiotherapy on pituitary corticotroph macrotumors in dogs: a retrospective study of 12 cases. Can Vet J 48: 481-486.
- Dillman R O. 2011. Cancer Immunotherapy. Cancer Biotherapy and Radiopharmaceuticals 26: 1-64.
- Disis M L, Bernhard H, Jaffee E M. 2009. Use of tumour-responsive T cells as cancer treatment. Lancet 373: 673-683.
- Finn O J. 2008. Cancer immunology. N Engl J Med 358: 2704-2715.
- Fridman W H, Pages F, Sautes-Fridman C, Galon J. 2012. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 12: 298-306.
- Hanahan D, Weinberg R A. 2011. Hallmarks of cancer: the next generation. Cell 144: 646-674.
- Jourdier T M, Moste C, Bonnet M C, Delisle F, Tafani J P, Devauchelle P, Tartaglia J, Moingeon P. 2003. Local immunotherapy of spontaneous feline fibrosarcomas using recombinant poxviruses expressing interleukin 2 (IL2). Gene Therapy 10: 2126-2132.
- Manley C A, Leibman N F, Wolchok J D, Riviere I C, Bartido S, Craft D M, Bergman P J. 2011. Xenogeneic murine tyrosinase DNA vaccine for malignant melanoma of the digit of dogs. J Vet Intern Med 25: 94-99.
- Marconato L. 2011. The staging and treatment of multicentric high-grade lymphoma in dogs: a review of recent developments and future prospects. Vet J 188: 34-38.
- Martinez P, Blasco M A. 2011. Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nature Reviews Cancer 11: 161-176.
- Mir L M. 2008. Application of electroporation gene therapy: past, current, and future. Methods Mol Biol 423: 3-17.
- Murray, 1991, ed. “Gene Transfer and Expression Protocols” Humana Press, Clifton, N.J.
- Sardesai N Y, Weiner D B. 2011. Electroporation delivery of DNA vaccines: prospects for success. Curr Opin Immunol 23: 421-429.
- Topalian S L, Weiner G J, Pardoll D M. 2011. Cancer Immunotherapy Comes of Age. Journal of Clinical Oncology 29: 4828-4836.
- Vascellari M, Baioni E, Ru G, Carminato A, Mutinelli F. 2009. Animal tumour registry of two provinces in northern Italy: incidence of spontaneous tumours in dogs and cats. BMC Vet Res 5: 39.
- Yang, 1992, “Gene transfer into mammalian somatic cells in vivo”, Crit. Rev. Biotech. 12: 335-356
- Yang Y, Chen Y, Zhang C, Huang H, Weissman S M. 2002. Nucleolar localization of hTERT protein is associated with telomerase function. Exp Cell Res 277: 201-209.
- Yazawa M, et al, 2003, J. Vet. Med. Sci 65(5):573-577